Characterization of intratumoral tertiary lymphoid structures in pancreatic ductal adenocarcinoma: cellular properties and prognostic significance

Background Tumor-associated tertiary lymphoid structures (TLSs) are functional immune-responsive niches that are not fully understood in pancreatic ductal adenocarcinoma (PDAC). Methods Fluorescent multiplex immunohistochemistry was performed on sequential sections of surgically resected tumor tissues from 380 PDAC patients without preoperative treatment (surgery alone (SA)) and 136 patients pretreated with neoadjuvant treatment (NAT). Multispectral images were processed via machine learning and image processing platforms, inForm V.2.4 and HALO V.3.2; TLS regions were segmented, and the cells were identified and quantified. The cellular composition and immunological properties of TLSs and their adjacent tissues in PDAC were scored and compared, and their association with prognosis was further examined. Results Intratumoral TLSs were identified in 21.1% (80/380) of patients in the SA group and 15.4% (21/136) of patients in the NAT group. In the SA group, the presence of intratumoral TLSs was significantly associated with improved overall survival (OS) and progression-free survival. The existence of intratumoral TLSs was correlated with elevated levels of infiltrating CD8+T, CD4+T, B cells and activated immune cells in adjacent tissues. A nomogram model was generated with TLS presence as a variable, which successfully predicted PDAC patient OS in an external validation cohort (n=123). In the NAT group, samples exhibited a lower proportion of B cells and a higher proportion of regulatory T cells within intratumoral TLSs. Additionally, these TLSs were smaller in size, with a lower overall maturation level and reduced immune cell activation, and the prognostic value of TLS presence was insignificant in the NAT cohort. Conclusion Our study systematically revealed the cellular properties and prognostic values of intratumoral TLSs in PDAC and described the potential impact of NAT on TLS development and function.

[1]  D. Olive,et al.  Regulatory T cells infiltrate the tumor-induced tertiary lymphoïd structures and are associated with poor clinical outcome in NSCLC , 2022, Communications biology.

[2]  J. Yun,et al.  Impact of mature tertiary lymphoid structures on prognosis and therapeutic response of Epstein-Barr virus-associated gastric cancer patients , 2022, Frontiers in Immunology.

[3]  O. Yazıcı,et al.  Tumor-Infiltrating Lymphocytes (TIL), Tertiary Lymphoid Structures (TLS), and Expression of PD-1, TIM-3, LAG-3 on TIL in Invasive and In Situ Ductal Breast Carcinomas and Their Relationship with Prognostic Factors. , 2022, Clinical breast cancer.

[4]  Erik N. Bergstrom,et al.  Mechanisms of APOBEC3 mutagenesis in human cancer cells , 2022, Nature.

[5]  T. Schumacher,et al.  Tertiary lymphoid structures in cancer , 2022, Science.

[6]  J. Yun,et al.  Distribution and density of tertiary lymphoid structures predict clinical outcome in intrahepatic cholangiocarcinoma. , 2021, Journal of hepatology.

[7]  Xueda Hu,et al.  Single-cell analyses reveal key immune cell subsets associated with response to PD-L1 blockade in triple-negative breast cancer. , 2021, Cancer cell.

[8]  Fangyu Xie,et al.  The OX40/OX40L Axis Regulates T Follicular Helper Cell Differentiation: Implications for Autoimmune Diseases , 2021, Frontiers in Immunology.

[9]  E. Nakakura,et al.  Pancreatic Adenocarcinoma, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. , 2021, Journal of the National Comprehensive Cancer Network : JNCCN.

[10]  E. Jaffee,et al.  Leukocyte Heterogeneity in Pancreatic Ductal Adenocarcinoma: Phenotypic and Spatial Features Associated with Clinical Outcome. , 2021, Cancer discovery.

[11]  D. Kwiatkowski,et al.  Tertiary lymphoid structure score: a promising approach to refine the TNM staging in resected non-small cell lung cancer , 2021, British Journal of Cancer.

[12]  A. Jemal,et al.  Cancer Statistics, 2021 , 2021, CA: a cancer journal for clinicians.

[13]  C. Dubay,et al.  Germinal center reactions in tertiary lymphoid structures associate with neoantigen burden, humoral immunity and long-term survivorship in pancreatic cancer , 2021, Oncoimmunology.

[14]  V. Engelhard,et al.  Insights into Tumor-Associated Tertiary Lymphoid Structures: Novel Targets for Antitumor Immunity and Cancer Immunotherapy , 2020, Cancer Immunology Research.

[15]  X. Lao,et al.  Worldwide Burden of, Risk Factors for, and Trends in Pancreatic Cancer. , 2020, Gastroenterology.

[16]  M. O'Hara,et al.  Challenges and Opportunities for Pancreatic Cancer Immunotherapy. , 2020, Cancer cell.

[17]  R. Kemp,et al.  Tertiary lymphoid structures in cancer – considerations for patient prognosis , 2020, Cellular & Molecular Immunology.

[18]  A. Maitra,et al.  Combination of PD1 Inhibitor and OX40 Agonist Induces Tumor Rejection and Immune Memory in Mouse Models of Pancreatic Cancer. , 2020, Gastroenterology.

[19]  D. Schadendorf,et al.  Tertiary lymphoid structures improve immunotherapy and survival in melanoma , 2020, Nature.

[20]  Anh Phong Tran,et al.  Delicate Balances in Cancer Chemotherapy: Modeling Immune Recruitment and Emergence of Systemic Drug Resistance , 2019, bioRxiv.

[21]  A. Krasinskas,et al.  Immunologic alterations in the pancreatic cancer microenvironment of patients treated with neoadjuvant chemo- and radiotherapy. , 2019, JCI insight.

[22]  H. Friess,et al.  Neoadjuvant Therapy Remodels the Pancreatic Cancer Microenvironment via Depletion of Protumorigenic Immune Cells , 2019, Clinical Cancer Research.

[23]  C. Sautès-Fridman,et al.  Tertiary lymphoid structures in the era of cancer immunotherapy , 2019, Nature Reviews Cancer.

[24]  T. Shichinohe,et al.  Prognostic relevance of tertiary lymphoid organs following neoadjuvant chemoradiotherapy in pancreatic ductal adenocarcinoma , 2019, Cancer science.

[25]  V. Janevska,et al.  Tertiary Lymphoid Structures in Colorectal Cancers and Their Prognostic Value , 2018, Open access Macedonian journal of medical sciences.

[26]  H. Lee,et al.  Presence of tertiary lymphoid structures determines the level of tumor-infiltrating lymphocytes in primary breast cancer and metastasis , 2018, Modern Pathology.

[27]  J. Kleeff,et al.  Therapeutic developments in pancreatic cancer: current and future perspectives , 2018, Nature Reviews Gastroenterology & Hepatology.

[28]  A. Gallimore,et al.  Tertiary Lymphoid Structures in Cancer: Drivers of Antitumor Immunity, Immunosuppression, or Bystander Sentinels in Disease? , 2017, Front. Immunol..

[29]  Mithat Gönen,et al.  Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer , 2017, Nature.

[30]  Nicole Joller,et al.  Regulatory T cells: balancing protection versus pathology. , 2016, Swiss medical weekly.

[31]  K. Horgan,et al.  Lymphocyte depletion and repopulation after chemotherapy for primary breast cancer , 2016, Breast Cancer Research.

[32]  Nikhil S. Joshi,et al.  Regulatory T Cells in Tumor-Associated Tertiary Lymphoid Structures Suppress Anti-tumor T Cell Responses. , 2015, Immunity.

[33]  Y. Kanai,et al.  Intratumoral tertiary lymphoid organ is a favourable prognosticator in patients with pancreatic cancer , 2015, British Journal of Cancer.

[34]  Mithat Gonen,et al.  Nomograms in oncology: more than meets the eye. , 2015, The Lancet. Oncology.

[35]  C. Sautès-Fridman,et al.  Tertiary lymphoid structures in cancer and beyond. , 2014, Trends in immunology.

[36]  M. Svoboda,et al.  B Cells and Ectopic Follicular Structures: Novel Players in Anti-Tumor Programming with Prognostic Power for Patients with Metastatic Colorectal Cancer , 2014, PloS one.

[37]  Youping Deng,et al.  Effect of Pemetrexed on Innate Immune Killer Cells and Adaptive Immune T Cells in Subjects With Adenocarcinoma of the Pancreas , 2012, Journal of immunotherapy.

[38]  Tibor Schuster,et al.  Preoperative/Neoadjuvant Therapy in Pancreatic Cancer: A Systematic Review and Meta-analysis of Response and Resection Percentages , 2010, PLoS medicine.

[39]  D. Kang,et al.  Significant Impairment in Immune Recovery After Cancer Treatment , 2009, Nursing research.

[40]  J. D. Capra,et al.  CD45RO enriches for activated, highly mutated human germinal center B cells. , 2007, Blood.

[41]  Holger Moch,et al.  Germinal Centers Determine the Prognostic Relevance of Tertiary Lymphoid Structures and Are Impaired by Corticosteroids in Lung Squamous Cell Carcinoma. , 2018, Cancer research.

[42]  L. Zitvogel,et al.  Rationale for anti-OX40 cancer immunotherapy. , 2016, European journal of cancer.